Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Med Chem ; 66(8): 5536-5549, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37057997

ABSTRACT

There is growing interest in non-psychoactive phytocannabinoids, namely cannabidiol (CBD), cannabigerol (CBG), and cannabichromene, as potential leads for novel therapeutic agents. In this study, we report on the development of new derivatives in which we methylated either position 4 of olivetol or the phenolic positions of olivetol, or both. We introduce a refinement on previously reported chemical procedures for phytocannabinoid derivatization as well as the biological evaluation of all derivatives in anti-inflammatory in vivo models. Compounds such as the CBD derivative, 2 and the CBG derivative, 11, significantly reduced cytokine levels when compared to their parent compounds. Moreover, both of these derivatives proved to be as potent as dexamethasone for the inhibition of IL-1ß. We believe that these new derivatives, as described herein, can be further developed as novel drug candidates for inflammatory conditions.


Subject(s)
Cannabidiol , Cannabidiol/pharmacology , Resorcinols , Anti-Inflammatory Agents , Cytokines
2.
Molecules ; 26(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34577072

ABSTRACT

Interest in CBG (cannabigerol) has been growing in the past few years, due to its anti-inflammatory properties and other therapeutic benefits. Here we report the synthesis of three new CBG derivatives (HUM-223, HUM-233 and HUM-234) and show them to possess anti-inflammatory and analgesic properties. In addition, unlike CBG, HUM-234 also prevents obesity in mice fed a high-fat diet (HFD). The metabolic state of the treated mice on HFD is significantly better than that of vehicle-treated mice, and their liver slices show significantly less steatosis than untreated HFD or CBG-treated ones from HFD mice. We believe that HUM-223, HUM-233 and HUM-234 have the potential for development as novel drug candidates for the treatment of inflammatory conditions, and in the case of HUM-234, potentially for obesity where there is a huge unmet need.


Subject(s)
Analgesics/chemical synthesis , Anti-Inflammatory Agents/chemical synthesis , Anti-Obesity Agents/chemical synthesis , Cannabinoids/chemistry , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Anti-Obesity Agents/therapeutic use , Fatty Liver/drug therapy , Female , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Osteoarthritis, Knee/drug therapy
3.
Proc Natl Acad Sci U S A ; 117(34): 20753-20763, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32759223

ABSTRACT

Fibrotic diseases remain a major cause of morbidity and mortality, yet there are few effective therapies. The underlying pathology of all fibrotic conditions is the activity of myofibroblasts. Using cells from freshly excised disease tissue from patients with Dupuytren's disease (DD), a localized fibrotic disorder of the palm, we sought to identify new therapeutic targets for fibrotic disease. We hypothesized that the persistent activity of myofibroblasts in fibrotic diseases might involve epigenetic modifications. Using a validated genetics-led target prioritization algorithm (Pi) of genome wide association studies (GWAS) data and a broad screen of epigenetic inhibitors, we found that the acetyltransferase CREBBP/EP300 is a major regulator of contractility and extracellular matrix production via control of H3K27 acetylation at the profibrotic genes, ACTA2 and COL1A1 Genomic analysis revealed that EP300 is highly enriched at enhancers associated with genes involved in multiple profibrotic pathways, and broad transcriptomic and proteomic profiling of CREBBP/EP300 inhibition by the chemical probe SGC-CBP30 identified collagen VI (Col VI) as a prominent downstream regulator of myofibroblast activity. Targeted Col VI knockdown results in significant decrease in profibrotic functions, including myofibroblast contractile force, extracellular matrix (ECM) production, chemotaxis, and wound healing. Further evidence for Col VI as a major determinant of fibrosis is its abundant expression within Dupuytren's nodules and also in the fibrotic foci of idiopathic pulmonary fibrosis (IPF). Thus, Col VI may represent a tractable therapeutic target across a range of fibrotic disorders.


Subject(s)
CREB-Binding Protein/genetics , Collagen Type VI/metabolism , E1A-Associated p300 Protein/metabolism , CREB-Binding Protein/metabolism , Cell Proliferation/drug effects , Collagen/metabolism , Collagen Type VI/physiology , E1A-Associated p300 Protein/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibrosis/genetics , Fibrosis/metabolism , Genome-Wide Association Study , Humans , Myofibroblasts/metabolism , Myofibroblasts/physiology , Proteomics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism
4.
Nat Commun ; 11(1): 2768, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488016

ABSTRACT

Fibrotic disorders are some of the most devastating and poorly treated conditions in developed nations, yet effective therapeutics are not identified for many of them. A major barrier for the identification of targets and successful clinical translation is a limited understanding of the human fibrotic microenvironment. Here, we construct a stromal cell atlas of human fibrosis at single cell resolution from patients with Dupuytren's disease, a localized fibrotic condition of the hand. A molecular taxonomy of the fibrotic milieu characterises functionally distinct stromal cell types and states, including a subset of immune regulatory ICAM1+ fibroblasts. In developing fibrosis, myofibroblasts exist along an activation continuum of phenotypically distinct populations. We also show that the tetraspanin CD82 regulates cell cycle progression and can be used as a cell surface marker of myofibroblasts. These findings have important implications for targeting core pathogenic drivers of human fibrosis.


Subject(s)
Dupuytren Contracture/immunology , Dupuytren Contracture/metabolism , Fibrosis/immunology , Fibrosis/metabolism , Stromal Cells/metabolism , Actins/metabolism , Biomarkers/metabolism , Chemokines, CXC/metabolism , Dupuytren Contracture/pathology , Fibrosis/pathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Molecular Medicine , Myofibroblasts/metabolism , Tetraspanins/metabolism , Tumor Microenvironment/physiology
5.
6.
Biol Open ; 9(3)2020 03 24.
Article in English | MEDLINE | ID: mdl-32139395

ABSTRACT

Mechanical force is a fundamental regulator of cell phenotype. Myofibroblasts are central mediators of fibrosis, a major unmet clinical need characterised by the deposition of excessive matrix proteins. Traction forces of myofibroblasts play a key role in remodelling the matrix and modulate the activities of embedded stromal cells. Here, we employ a combination of unsupervised computational analysis, cytoskeletal profiling and single cell traction force microscopy as a functional readout to uncover how the complex spatiotemporal dynamics and mechanics of living human myofibroblast shape sub-cellular profiling of traction forces in fibrosis. We resolve distinct biophysical communities of myofibroblasts, and our results provide a new paradigm for studying functional heterogeneity in human stromal cells.


Subject(s)
Biophysical Phenomena , Myofibroblasts/physiology , Single-Cell Analysis , Biomarkers , Biomechanical Phenomena , Cells, Cultured , Cytoskeleton/metabolism , Fluorescent Antibody Technique , Humans , Molecular Imaging , Myofibroblasts/cytology , Single-Cell Analysis/methods
7.
Sci Adv ; 5(12): eaay0370, 2019 12.
Article in English | MEDLINE | ID: mdl-31840071

ABSTRACT

Dissecting the molecular landscape of fibrotic disease, a major unmet need, will inform the development of novel treatment strategies to target disease progression and identify desperately needed therapeutic targets. Here, we provide a detailed single-cell analysis of the immune landscape in Dupuytren's disease, a localized fibrotic condition of the hand, and identify a pathogenic signaling circuit between stromal and immune cells. We demonstrate M2 macrophages and mast cells as key cellular sources of tumor necrosis factor (TNF) that promotes myofibroblast development. TNF acts via the inducible TNFR2 receptor and stimulates interleukin-33 (IL-33) secretion by myofibroblasts. In turn, stromal cell IL-33 acts as a potent stimulus for TNF production from immune cells. Targeting this reciprocal signaling pathway represents a novel therapeutic strategy to inhibit the low-grade inflammation in fibrosis and the mechanism that drives chronicity.


Subject(s)
Dupuytren Contracture/genetics , Fibrosis/genetics , Interleukin-33/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Tumor Necrosis Factor-alpha/genetics , Cell Line , Dupuytren Contracture/drug therapy , Dupuytren Contracture/immunology , Dupuytren Contracture/pathology , Fibrosis/drug therapy , Fibrosis/immunology , Fibrosis/pathology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/pathology , Molecular Targeted Therapy , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction/genetics , Single-Cell Analysis/methods , Tumor Necrosis Factor-alpha/immunology
8.
Proc Natl Acad Sci U S A ; 116(43): 21666-21672, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31597740

ABSTRACT

Regulatory T (Treg) cells expressing the transcription factor Foxp3 play an important role in maintaining immune homeostasis. Chronic inflammation is associated with reduced Foxp3 expression, function, and loss of phenotypic stability. Previous studies have established the importance of TNF receptor 2 (TNFR2) in the generation and/or activation of Treg cells. In this study, we assess the importance of TNFR2 in healthy mice and under inflammatory conditions. Our findings reveal that, in health, TNFR2 is important not only for the generation of Treg cells, but also for regulating their functional activity. We also show that TNFR2 maintains Foxp3 expression in Treg cells by restricting DNA methylation at the Foxp3 promoter. In inflammation, loss of TNFR2 results in increased severity and chronicity of experimental arthritis, reduced total numbers of Treg cells, reduced accumulation of Treg cells in inflamed joints, and loss of inhibitory activity. In addition, we demonstrate that, under inflammatory conditions, loss of TNFR2 causes Treg cells to adopt a proinflammatory Th17-like phenotype. It was concluded that TNFR2 signaling is required to enable Treg cells to promote resolution of inflammation and prevent them from undergoing dedifferentiation. Consequently, TNFR2-specific agonists or TNF1-specific antagonists may be useful in the treatment of autoimmune disease.


Subject(s)
Autoimmune Diseases/immunology , DNA Methylation/genetics , Forkhead Transcription Factors/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Promoter Regions, Genetic/genetics
9.
Nat Genet ; 51(7): 1082-1091, 2019 07.
Article in English | MEDLINE | ID: mdl-31253980

ABSTRACT

Most candidate drugs currently fail later-stage clinical trials, largely due to poor prediction of efficacy on early target selection1. Drug targets with genetic support are more likely to be therapeutically valid2,3, but the translational use of genome-scale data such as from genome-wide association studies for drug target discovery in complex diseases remains challenging4-6. Here, we show that integration of functional genomic and immune-related annotations, together with knowledge of network connectivity, maximizes the informativeness of genetics for target validation, defining the target prioritization landscape for 30 immune traits at the gene and pathway level. We demonstrate how our genetics-led drug target prioritization approach (the priority index) successfully identifies current therapeutics, predicts activity in high-throughput cellular screens (including L1000, CRISPR, mutagenesis and patient-derived cell assays), enables prioritization of under-explored targets and allows for determination of target-level trait relationships. The priority index is an open-access, scalable system accelerating early-stage drug target selection for immune-mediated disease.


Subject(s)
Arthritis, Rheumatoid/genetics , Drug Discovery , Gene Regulatory Networks , Genome, Human , Immunity, Innate/genetics , Quantitative Trait Loci , Selection, Genetic , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Gene Expression Regulation , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide
10.
Methods Enzymol ; 610: 27-58, 2018.
Article in English | MEDLINE | ID: mdl-30390803

ABSTRACT

Chemical probes are small molecules with potency and selectivity for a single or small number of protein targets. A good chemical probe engages its target intracellularly and is accompanied by a chemically similar, but inactive molecule to be used as a negative control in cellular phenotypic screening. The utility of these chemical probes is ultimately governed by how well they are developed and characterized. Chemical probes either as single entities, or in chemical probes sets are being increasingly used to interrogate the biological relevance of a target in a disease model. This chapter lays out the core properties of chemical probes, summarizes the seminal and emerging techniques used to demonstrate robust intracellular target engagement. Translation of target engagement assays to disease-relevant phenotypic assays using primary patient-derived cells and tissues is also reviewed. Two examples of epigenetic chemical probe discovery and utility are presented whereby target engagement pointed to novel disease associations elucidated from poorly understood protein targets. Finally, a number of examples are discussed whereby chemical probe sets, or "chemogenomic libraries" are used to illuminate new target-disease links which may represent future directions for chemical probe utility.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Epigenesis, Genetic/drug effects , Humans , Molecular Targeted Therapy/methods
11.
EBioMedicine ; 33: 282-288, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29983350

ABSTRACT

BACKGROUND: Dupuytren's disease is a common fibrotic condition of the hand that causes irreversible flexion contractures of the fingers, with no approved therapy for early stage disease. Our previous analysis of surgically-excised tissue defined tumour necrosis factor (TNF) as a potential therapeutic target. Here we assessed the efficacy of injecting nodules of Dupuytren's disease with a TNF inhibitor. METHODS: Patients were randomised to receive adalimumab on one occasion in dose cohorts of 15 mg in 0.3 ml, 35 mg in 0.7 ml, or 40 mg in 0.4 ml, or an equivalent volume of placebo in a 3:1 ratio. Two weeks later the injected tissue was surgically excised and analysed. The primary outcome measure was levels of mRNA expression for α-smooth muscle actin (ACTA2). Secondary outcomes included levels of α-SMA and collagen proteins. The trial was registered with ClinicalTrial.gov (NCT03180957) and the EudraCT (2015-001780-40). FINDINGS: We recruited 28 patients, 8 assigned to the 15 mg, 12 to the 35 mg and 8 to the 40 mg adalimumab cohorts. There was no change in mRNA levels for ACTA2, COL1A1, COL3A1 and CDH11. Levels of α-SMA protein expression in patients treated with 40 mg adalimumab (1.09 ±â€¯0.09 ng per µg of total protein) were significantly lower (p = 0.006) compared to placebo treated patients (1.51 ±â€¯0.09 ng/µg). The levels of procollagen type I protein expression were also significantly lower (p < 0.019) in the sub group treated with 40 mg adalimumab (474 ±â€¯84 pg/µg total protein) compared with placebo (817 ±â€¯78 pg/µg). There were two serious adverse events, both considered unrelated to the study drug. INTERPRETATION: In this dose-ranging study, injection of 40 mg of adalimumab in 0.4 ml resulted in down regulation of the myofibroblast phenotype as evidenced by reduction in expression of α-SMA and type I procollagen proteins at 2 weeks. These data form the basis of an ongoing phase 2b clinical trial assessing the efficacy of intranodular injection of 40 mg adalimumab in 0.4 ml compared to an equivalent volume of placebo in patients with early stage Dupuytren's disease. FUNDING: Health Innovation Challenge Fund (Wellcome Trust and Department of Health) and 180 Therapeutics LP.


Subject(s)
Actins/metabolism , Adalimumab/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Collagen Type I/metabolism , Dupuytren Contracture/drug therapy , Actins/genetics , Adalimumab/pharmacology , Anti-Inflammatory Agents/pharmacology , Collagen Type I/genetics , Double-Blind Method , Down-Regulation , Drug Administration Schedule , Dupuytren Contracture/genetics , Dupuytren Contracture/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Injections , Male , Treatment Outcome
12.
Cytokine ; 101: 19-25, 2018 Jan.
Article in English | MEDLINE | ID: mdl-33730773

ABSTRACT

Tumour necrosis factor-α (TNF-α) is a highly pleiotropic cytokine with effects on multiple pathological and physiological functions via two distinct receptors, TNFR1 and TNFR2. Much of the pro- inflammatory action of TNF-α is mediated by TNFR1 whereas TNFR2 is thought to play an immunoregulatory and tissue protective role. Anti-TNF- α biologics have been extremely successful in treating a number of immune mediated pathologies, including rheumatoid arthritis, ankylosing spondylitis, psoriasis, psoriatic arthritis and inflammatory bowel disease. However, anti-TNF therapy has been shown to induce systemic lupus erythematosus and psoriasis in some patients, and to be deleterious in multiple sclerosis. It is hypothesized that these paradoxical effects of anti-TNF-α are due to inhibition of TNFR2 signalling. In this review, we will focus on the biology and pathophysiologic role of TNF-α and on the therapeutic implications of targeting TNF-α receptor signalling.

13.
Nat Commun ; 8(1): 1595, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29150600

ABSTRACT

Pattern recognition underpins innate immunity; the accurate identification of danger, including infection, injury, or tumor, is key to an appropriately targeted immune response. Pathogen detection is increasingly well defined mechanistically, but the discrimination of endogenous inflammatory triggers remains unclear. Tenascin-C, a matrix protein induced upon tissue damage and expressed by tumors, activates toll-like receptor 4 (TLR4)-mediated sterile inflammation. Here we map three sites within tenascin-C that directly and cooperatively interact with TLR4. We also identify a conserved inflammatory epitope in related proteins from diverse families, and demonstrate that its presence targets molecules for TLR detection, while its absence enables escape of innate immune surveillance. These data reveal a unique molecular code that defines endogenous proteins as inflammatory stimuli by marking them for recognition by TLRs.


Subject(s)
Immunity, Innate , Inflammation/metabolism , Tenascin/metabolism , Toll-Like Receptor 4/metabolism , Amino Acid Sequence , Binding Sites/genetics , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Interaction Mapping , Sequence Homology, Amino Acid , Signal Transduction , Tenascin/chemistry , Tenascin/genetics , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/genetics
14.
Methods Mol Biol ; 1390: 351-81, 2016.
Article in English | MEDLINE | ID: mdl-26803640

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent synovial inflammation leading to tissue destruction and progressive loss of joint function. Here we describe two methods that can be used to assess the contribution of toll-like receptors (TLRs), and their potential ligands, to RA pathogenesis. We focus on the antigen-induced model of murine arthritis and human synovial tissue explant models. Both enable detection of TLR, and TLR ligand, expression, as well as investigation of the effect of inhibition of these molecules. Each offers a unique insight into disease; with murine models allowing kinetic analysis in live animals and explant models allowing examination of inflamed human tissue, which together can help us to dissect the role of TLRs in the onset and progression of RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , Toll-Like Receptors/metabolism , Animals , Antigens/immunology , Cell Culture Techniques , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Mice , Phenotype , Real-Time Polymerase Chain Reaction , Synovial Membrane/immunology , Synovial Membrane/metabolism , Toll-Like Receptors/genetics
15.
J Exp Med ; 212(6): 845-53, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25941255

ABSTRACT

The most studied biological role of type III interferons (IFNs) has so far been their antiviral activity, but their role in autoimmune and inflammatory diseases remains largely unexplored. Here, we show that treatment with IFN-λ2/IL-28A completely halts and reverses the development of collagen-induced arthritis (CIA) and discover cellular and molecular mechanisms of IL-28A antiinflammatory function. We demonstrate that treatment with IL-28A dramatically reduces numbers of proinflammatory IL-17-producing Th17 and γδ T cells in the joints and inguinal lymph nodes, without affecting T cell proliferative responses or levels of anticollagen antibodies. IL-28A exerts its antiinflammatory effect by restricting recruitment of IL-1b-expressing neutrophils, which are important for amplification of inflammation. We identify neutrophils as cells expressing high levels of IFN-λ receptor 1 (IFNLR1)-IL-28 receptor α (IL28RA) and targeted by IL-28A. Our data highlight neutrophils as contributors to the pathogenesis of autoimmune arthritis and present IFN-λs or agonists of IFNLR1-IL28RA as putative new therapeutics for neutrophil-driven inflammation.


Subject(s)
Gene Expression Regulation , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukins/pharmacology , Animals , Arthritis/metabolism , CD4-Positive T-Lymphocytes/cytology , Cattle , Cell Movement , Cell Proliferation , Chickens , Collagen/chemistry , Flow Cytometry , Interleukin-17/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neutrophils/cytology , Neutrophils/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Cytokine/metabolism , T-Lymphocytes/cytology
16.
Arthritis Rheumatol ; 66(10): 2728-38, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24965881

ABSTRACT

OBJECTIVE: Tumor necrosis factor (TNF) signals via 2 receptors, TNFR type I (TNFRI) and TNFRII, with distinct cellular distribution and signaling functions. In rheumatoid arthritis (RA), the net effect of TNFR signaling favors inflammatory responses while inhibiting the activity of regulatory T cells. TNFRII signaling has been shown to promote Treg cell function. To assess the relative contributions of TNFRI and TNFRII signaling to inflammatory and regulatory responses in vivo, we compared the effect of TNF blockade, hence TNFRI/II, versus TNFRI alone in collagen-induced arthritis (CIA) as a model of RA. METHODS: Mice with established arthritis were treated for 10 days with anti-mouse TNFRI domain antibody (dAb; DMS5540), an isotype control dAb (DMS5538), or murine TNFRII genetically fused with mouse IgG1 Fc domain (mTNFRII-Fc) beginning on the day of arthritis onset, and disease progression was monitored. Systemic cytokine concentrations and numbers of T cell subsets in lymph nodes and spleens were measured, and intrinsic Treg cell function was determined by ex vivo suppression assays. RESULTS: Progression of CIA was suppressed similarly by TNFRI (DMS5540) and TNFRI/II (mTNFRII-Fc) blockade. However, blockade of TNFRI/II led to increased effector T cell activity, which was not observed after selective TNFRI blockade, suggesting an immunoregulatory role of TNFRII. In support of this, TNFRI blockade, but not TNFRI/II blockade, expanded and activated Treg cells. Furthermore, a dramatic increase in expression of the Treg cell signature genes FoxP3 and TNFRII was observed in joints undergoing remission, which supports the notion that these molecules have a physiologic role in the resolution of inflammation. CONCLUSION: We propose that a therapeutic strategy that targets TNFRI while sparing TNFRII has the potential to both inhibit inflammation and promote Treg cell activity, which might be superior to TNF blockade.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Single-Domain Antibodies/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Cell Proliferation/drug effects , Forkhead Transcription Factors/metabolism , Inflammation/drug therapy , Inflammation/immunology , Male , Mice , Mice, Inbred DBA , Recombinant Fusion Proteins/pharmacology , Single-Domain Antibodies/pharmacology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/immunology
17.
Proc Natl Acad Sci U S A ; 111(6): 2289-94, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469824

ABSTRACT

An immune response is essential for protection against infection, but, in many individuals, aberrant responses against self tissues cause autoimmune diseases such as rheumatoid arthritis (RA). How to diminish the autoimmune response while not augmenting infectious risk is a challenge. Modern targeted therapies such as anti-TNF or anti-CD20 antibodies ameliorate disease, but at the cost of some increase in infectious risk. Approaches that might specifically reduce autoimmunity and tissue damage without infectious risk would be important. Here we describe that TNF superfamily member OX40 ligand (OX40L; CD252), which is expressed predominantly on antigen-presenting cells, and its receptor OX40 (on activated T cells), are restricted to the inflamed joint in arthritis in mice with collagen-induced arthritis and humans with RA. Blockade of this pathway in arthritic mice reduced inflammation and restored tissue integrity predominantly by inhibiting inflammatory cytokine production by OX40L-expressing macrophages. Furthermore, we identify a previously unknown role for OX40L in steady-state bone homeostasis. This work shows that more targeted approaches may augment the "therapeutic window" and increase the benefit/risk in RA, and possibly other autoimmune diseases, and are thus worth testing in humans.


Subject(s)
Arthritis, Rheumatoid/therapy , Membrane Glycoproteins/immunology , Osteoclasts/cytology , Tumor Necrosis Factors/immunology , Animals , Antibodies, Monoclonal/immunology , Arthritis, Rheumatoid/pathology , Cytokines/biosynthesis , Homeostasis , Inflammation Mediators/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Mice , OX40 Ligand , Signal Transduction , Tumor Necrosis Factor Inhibitors
18.
Cell Signal ; 26(4): 683-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24378531

ABSTRACT

Tumour necrosis factor (p55 or p60) receptor (TNFR) 1 is the major receptor that activates pro-inflammatory signalling and induces gene expression in response to TNF. Consensus is lacking for the function of (p75 or p80) TNFR2 but experiments in mice have suggested neuro-, cardio- and osteo-protective and anti-inflammatory roles. It has been shown in various cell types to be specifically required for the induction of TNFR-associated factor-2 (TRAF2) degradation and activation of the alternative nuclear factor (NF)-kappaB pathway, and to contribute to the activation of mitogen-activated protein kinases (MAPK) and the classical NF-kappaB pathway. We have investigated the signalling functions of TNFR2 in primary human and murine macrophages. We find that in these cells TNF induces TRAF2 degradation, and this is blocked in TNFR2(-/-) macrophages. TRAF2 has been previously reported to be required for TNF-induced activation of p38 MAPK. However, TRAF2 degradation does not inhibit TNF-induced tolerance of p38 MAPK activation. Neither TNF, nor lipopolysaccharide treatment, induced activation of the alternative NF-kappaB pathway in macrophages. Activation by TNF of the p38 MAPK and NF-kappaB pathways was blocked in TNFR1(-/-) macrophages. In contrast, although TNFR2(-/-) macrophages displayed robust p38 MAPK activation and IkappaBα degradation at high concentrations of TNF, at lower doses the concentration dependence of signalling was weakened by an order of magnitude. Our results suggest that, in addition to inducing TRAF2 protein degradation, TNFR2 also plays a crucial auxiliary role to TNFR1 in sensitising macrophages for the ligand-induced activation of the p38 MAPK and classical NF-kappaB pro-inflammatory signalling pathways.


Subject(s)
Macrophages/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , TNF Receptor-Associated Factor 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Humans , I-kappa B Proteins/metabolism , Ligands , Lipopolysaccharides/pharmacology , Macrophages/cytology , Mice , NF-KappaB Inhibitor alpha , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
19.
Drug Des Devel Ther ; 7: 201-10, 2013.
Article in English | MEDLINE | ID: mdl-23569359

ABSTRACT

Psoriasis is a chronic inflammatory skin disease, most commonly resulting in the occurrence of red and silver scaly plaques. About 30% of psoriasis sufferers develop psoriatic arthritis (PsA), a disorder that presents with additional joint inflammation and other clinical features. At present, the most effective treatment for moderate and severe psoriasis and PsA are biologics such as antitumor necrosis factor alpha therapy. Biologics are costly and typically require repeated injections; hence, the development of novel, orally available, small molecular inhibitors that are less expensive to produce is highly desirable. The phosphodiesterase 4 inhibitor apremilast is a small molecular inhibitor that acts by increasing cyclic adenosine monophosphate levels, ultimately suppressing tumor necrosis alpha production. Apremilast has been tested in a number of psoriasis and PsA pilot and Phase II trials to evaluate its efficacy and safety. More recently, three larger double-blinded, and randomized multicenter studies demonstrate that apremilast is efficacious in the treatment of psoriasis and PsA, with significantly higher numbers of apremilast-treated patients achieving endpoints of a 75% reduction compared to baseline in Psoriasis Area and Severity Index (PASI-75) or American College of Rheumatology-20 scores, relative to placebo. This encouraging data, along with a tolerable incidence of mild to moderate adverse events, has led to the initiation of several large Phase III trials that aim to further validate apremilast as a treatment for psoriasis and PsA. Here, we provide an overview of the current treatments for psoriasis and PsA, and summarize the findings from multiple Phase II clinical trials where the effects of apremilast in the treatment of psoriasis and PsA patients have been investigated.


Subject(s)
Arthritis, Psoriatic/drug therapy , Psoriasis/drug therapy , Thalidomide/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arthritis, Psoriatic/physiopathology , Clinical Trials, Phase II as Topic , Humans , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Psoriasis/pathology , Severity of Illness Index , Thalidomide/adverse effects , Thalidomide/pharmacology , Thalidomide/therapeutic use
20.
PLoS One ; 7(12): e49894, 2012.
Article in English | MEDLINE | ID: mdl-23251353

ABSTRACT

UNLABELLED: Procalcitonin has been shown to be useful in separating infection from non-infective disorders. However, infection is often paralleled by tissue inflammation. Most studies supporting the use of procalcitonin were confounded by more significant inflammation in the infection group. Few studies have examined the usefulness of procalcitonin when adjusted for inflammation.Pleural inflammation underlies the development of most exudative effusions including pleural infection and malignancy. Pleurodesis, often used to treat effusions, involves provocation of intense aseptic pleural inflammation. We conducted a two-part proof-of-concept study to test the specificity of procalcitonin in differentiating infection using cohorts of patients with pleural effusions of infective and non-infective etiologies, as well as subjects undergoing pleurodesis. METHODS: We measured the blood procalcitonin level (i) in 248 patients with pleural infection or with non-infective pleural inflammation, matched for severity of systemic inflammation by C-reactive protein (CRP), age and gender; and (ii) in patients before and 24-48 hours after induction of non-infective pleural inflammation (from talc pleurodesis). RESULTS: 1) Procalcitonin was significantly higher in patients with pleural infection compared with controls with non-infective effusions (n = 32 each group) that were case-matched for systemic inflammation as measured by CRP [median (25-75%IQR): 0.58 (0.35-1.50) vs 0.34 (0.31-0.42) µg/L respectively, p = 0.003]. 2) Talc pleurodesis provoked intense systemic inflammation, and raised serum CRP by 360% over baseline. However procalcitonin remained relatively unaffected (21% rise). 3) Procalcitonin and CRP levels did not correlate. In 214 patients with pleural infection, procalcitonin levels did not predict the survival or need for surgical intervention. CONCLUSION: Using a pleural model, this proof-of-principle study confirmed that procalcitonin is a biomarker specific for infection and is not affected by non-infective inflammation. Procalcitonin is superior to CRP in distinguishing infection from non-infective pleural diseases, even when controlled for the level of systemic inflammation.


Subject(s)
Calcitonin/blood , Infections/diagnosis , Inflammation/diagnosis , Pleural Diseases/diagnosis , Protein Precursors/blood , Aged , Aged, 80 and over , Biomarkers/blood , Calcitonin Gene-Related Peptide , Diagnosis, Differential , Female , Humans , Infections/blood , Inflammation/blood , Male , Middle Aged , Pleura/metabolism , Pleural Diseases/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...